Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Steroids ; 97: 2-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25065587

ABSTRACT

The skin irritating principle from Thapsia garganica was isolated, named thapsigargin and the structure elucidated. By inhibiting the sarco/endoplasmic reticulum Ca(2+) ATPase (SERCA) thapsigargin provokes apoptosis in almost all cells. By conjugating thapsigargin to peptides, which are only substrates for either prostate specific antigen (PSA) or prostate specific membrane antigen (PSMA) prodrugs were created, which selectively affect prostate cancer cells or neovascular tissue in tumors. One of the prodrug is currently tested in clinical phase II. The prodrug under clinical trial has been named mipsagargin.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apiaceae/chemistry , Enzyme Inhibitors/pharmacology , Prodrugs/pharmacology , Prostatic Neoplasms/drug therapy , Soft Tissue Neoplasms/drug therapy , Thapsigargin/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Humans , Male , Mice , Molecular Structure , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Prodrugs/chemistry , Prodrugs/isolation & purification , Prostatic Neoplasms/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Soft Tissue Neoplasms/pathology , Thapsigargin/chemistry , Thapsigargin/isolation & purification
2.
Sci Transl Med ; 4(140): 140ra86, 2012 Jun 27.
Article in English | MEDLINE | ID: mdl-22745436

ABSTRACT

Heterogeneous expression of drug target proteins within tumor sites is a major mechanism of resistance to anticancer therapies. We describe a strategy to selectively inhibit, within tumor sites, the function of a critical intracellular protein, the sarcoplasmic/endoplasmic reticulum calcium adenosine triphosphatase (SERCA) pump, whose proper function is required by all cell types for viability. To achieve targeted inhibition, we took advantage of the unique expression of the carboxypeptidase prostate-specific membrane antigen (PSMA) by tumor endothelial cells within the microenvironment of solid tumors. We generated a prodrug, G202, consisting of a PSMA-specific peptide coupled to an analog of the potent SERCA pump inhibitor thapsigargin. G202 produced substantial tumor regression against a panel of human cancer xenografts in vivo at doses that were minimally toxic to the host. On the basis of these data, a phase 1 dose-escalation clinical trial has been initiated with G202 in patients with advanced cancer.


Subject(s)
Antigens, Surface/metabolism , Endothelial Cells/metabolism , Glutamate Carboxypeptidase II/metabolism , Prodrugs/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , Cell Line, Tumor , Humans , Male , Mice
3.
Anticancer Agents Med Chem ; 9(3): 276-94, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19275521

ABSTRACT

Available chemotherapeutics take advantage of the fast proliferation of cancer cells. Consequently slow growth makes androgen refractory prostate cancer resistant towards available drugs. No treatment is available at the present, when the cancer has developed metastases outside the prostate (T4 stage). Cytotoxins killing cells irrespective of the phase of the cell cycle will be able to kill slowly proliferating prostate cancer cells. Lack of selectivity, however, prevents their use as systemic drugs. Prostate cancer cells secrete characteristic proteolytic enzymes, e.g. PSA and hK2, with unusual substrate specificity. Conjugation of cytotoxins with peptides, which are selective substrates for PSA or hK2, will afford prodrugs, from which the active drug only will be released in close vicinity of the cancer cells. Based on this strategy prodrugs targeted at prostate cancer cells have been constructed and evaluated as potential drugs for prostate cancer. The potency of the thapsigargins as apoptotic agents make these naturally occurring sesquiterpene lactones attractive lead compounds. Intensive studies on structure-activity relationships and chemistry of the thapsigargins have enabled construction of potent derivatives enabling conjugation with peptides. Studies on the mechanism of action of the thapsigargins have revealed that the cytoxicity is based on their ability to inhibit the intracellular sarco-/endoplasmtic calcium pump.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Drug Design , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Thapsigargin/pharmacology , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Male , Prostatic Neoplasms/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Structure-Activity Relationship , Thapsigargin/chemical synthesis , Thapsigargin/chemistry
4.
Prostate ; 66(4): 358-68, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16302271

ABSTRACT

BACKGROUND: Prostate cancer cells secrete unique proteases such as prostate-specific antigen (PSA) and human glandular kallikrein 2 (hK2) that represent targets for the activation of prodrugs as systemic treatment of metastatic prostate cancer. Previously, a combinatorial peptide library was screened to identify a highly active peptide substrate for hK2. The peptide was coupled to an analog of the potent cytotoxin thapsigargin, L12ADT, to generate an hK2-activated prodrug that was efficiently hydrolyzed by purified hK2, stable to hydrolysis in human and mouse plasma in vitro and selectively toxic to hK2 producing prostate cancer cells in vitro. METHODS: In the current study, toxicology, pharmacokinetics, prodrug biodistribution, and antitumor efficacy studies were performed to evaluate the hK2-activated prodrug in vivo. RESULTS: The single intravenous maximally tolerated dose of prodrug was 6 mg/kg (i.e., 3.67 micromole/kg) which produced peak serum concentration of approximately 36 microM and had a half-life of approximately 40 min. In addition, over a 24 hr period <0.5% of free L12ADT analog was observed in plasma. The prodrug demonstrated significant antitumor effect in vivo while it was being administered, but prolonged intravenous administration was not possible due to local toxicity to tail veins. Subcutaneous administration of equimolar doses produced lower plasma AUC compared to intravenous dosing but equivalent intratumoral levels of prodrug following multiple doses. CONCLUSIONS: The hK2-activated prodrug was stable in vivo. The prodrug, however, was rapidly cleared and difficult to administer over prolonged dosing interval. Additional studies are underway to assess antitumor efficacy with prolonged administration of higher subcutaneous doses of prodrug. Second-generation hK2-activated thapsigargin prodrugs with increased half-lives and improved formulations are also under development.


Subject(s)
Prodrugs/pharmacology , Prodrugs/pharmacokinetics , Prostatic Neoplasms/pathology , Thapsigargin/pharmacology , Thapsigargin/pharmacokinetics , Tissue Kallikreins/pharmacology , Tissue Kallikreins/pharmacokinetics , Animals , Area Under Curve , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Mice , Mice, Inbred BALB C , Thapsigargin/analogs & derivatives , Tumor Cells, Cultured
5.
J Med Chem ; 46(25): 5375-88, 2003 Dec 04.
Article in English | MEDLINE | ID: mdl-14640546

ABSTRACT

A series of potent vascular endothelial growth factor R2 (VEGF-R2) tyrosine kinase inhibitors from a new indenopyrrolocarbazole template is reported. The structure-activity relationships for a series of 9-alkoxymethyl-12-(3-hydroxypropyl)indeno[2,1-a]pyrrolo[3,4-c]carbazole-5-ones revealed an optimal R9 substitution with ethoxymethyl 19 (VEGF-R2 IC(50) = 4 nM) and isopropoxymethyl 21 (VEGF-R2 IC(50) = 8 nM) being the most potent inhibitors in the series. The VEGF-R2 activity was reduced appreciably by increasing the size of the R9 alkoxy group or by alpha-methyl branching adjacent to the ring. The combined R9 alkoxymethyl and N12 hydroxypropyl substitutions were required for potent VEGF-R2 activity, and the corresponding thioether analogues were weaker than their ether counterparts. Compound 21 (R9 isopropoxymethyl, CEP-5214) was identified as a potent, low-nanomolar pan inhibitor of human VEGF-R tyrosine kinases, displaying IC(50) values of 16, 8, and 4 nM for VEGF-R1/FLT-1, VEGF-R2/KDR, and VEGF-R3/FLT-4, respectively, with cellular activity equivalent to the isolated enzyme activity. Compound 21 exhibited good selectivity against numerous tyrosine and serine/threonine kinases including PKC, Tie2, TrkA, CDK1, p38, JNK, and IRK. To increase water solubility and oral bioavailability, the N,N-dimethylglycine ester 40 was prepared. In pharmacokinetic studies in mice and rats, increased plasma levels of 21 were observed after oral administration of 40. Compound 21 demonstrated significant in vivo antitumor activity in numerous tumor models and was advanced into phase I clinical trials as the water-soluble N,N-dimethylglycine ester prodrug 40 (CEP-7055).


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Carbazoles/chemical synthesis , Prodrugs/chemical synthesis , Sarcosine/analogs & derivatives , Sarcosine/chemistry , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Carbazoles/chemistry , Carbazoles/pharmacology , Cells, Cultured , Drug Screening Assays, Antitumor , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Enzyme-Linked Immunosorbent Assay , Female , Hemangiosarcoma/drug therapy , Humans , In Vitro Techniques , Indenes/chemical synthesis , Indenes/chemistry , Indenes/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Nude , Models, Molecular , Phosphorylation , Prodrugs/chemistry , Prodrugs/pharmacology , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Solubility , Structure-Activity Relationship
6.
Cancer Res ; 63(18): 5978-91, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-14522925

ABSTRACT

Inhibition of the vascular endothelial growth factor VEGF-VEGF receptor (VEGF-R) kinase axes in the tumor angiogenic cascade is a promising therapeutic strategy in oncology. CEP-7055 is the fully synthetic orally active N,N-dimethyl glycine ester of CEP-5214, a C3-(isopropylmethoxy) fused pyrrolocarbazole with potent pan-VEGF-R kinase inhibitory activity. CEP-5214 demonstrates IC(50) values of 18 nM, 12 nM, and 17 nM against human VEGF-R2/KDR kinase, VEGF-R1/FLT-1 kinase, and VEGF-R3/FLT-4 kinase, respectively, in biochemical kinase assays. CEP-5214 inhibited VEGF-stimulated VEGF-R2/KDR autophosphorylation in human umbilical vein endothelial cells (HUVECs) with an IC (50) of approximately 10 nM and demonstrated an equivalent inhibition of murine FLK-1 autophosphorylation in transformed SVR endothelial cells. Evaluation of the antiangiogenic activity of CEP-5214 revealed a dose-related inhibition of microvessel growth ex vivo in rat aortic ring explant cultures and in vitro on HUVEC capillary-tube formation on Matrigel at low nanomolar concentrations. The antiangiogenic activity of CEP-5214 in these bioassays was observed in the absence of apparent cytotoxicity. Single-dose p.o. or s.c. administration of CEP-7055 or CEP-5214 to CD-1 mice at 23.8 mg/kg/dose b.i.d. resulted in a reversible inhibition of VEGF-R2/FLK-1 phosphorylation in murine lung tissues. Administration p.o. of CEP-7055 at 2.57 to 23.8 mg/kg/dose b.i.d. resulted in dose-related reductions in neovascularization in vivo in porcine aortic endothelial cell (PAEC)-VEGF/basic fibroblast growth factor-Matrigel implants in nude mice (maximum, 82% inhibition), significant reductions in granuloma formation (30%) and granuloma vascularity (42%) in a murine chronic inflammation-induced angiogenesis model, and significant and sustained (6 h) inhibition of VEGF-induced plasma extravasation in rats, with an ED(50) of 20 mg/kg/dose. Chronic p.o. administration of CEP-7055 at doses of 11.9 to 23.8 mg/kg/dose b.i.d. resulted in significant inhibition (50-90% maximum inhibition relative to controls) in the growth of a variety of established murine and human s.c. tumor xenografts in nude mice, including A375 melanomas, U251MG and U87MG glioblastomas, CALU-6 lung carcinoma, ASPC-1 pancreatic carcinoma, HT-29 and HCT-116 colon carcinomas, MCF-7 breast carcinomas, and SVR angiosarcomas. Significant antitumor efficacy was observed similarly against orthotopically implanted LNCaP human prostate carcinomas in male nude mice and orthotopically implanted renal carcinoma (RENCA) tumors in BALB/c mice, in terms of a significant reduction in the metastatic score and the extent of pulmonary metastases. These antitumor responses were associated with marked increases in tumor apoptosis, and significant reductions in intratumoral microvessel density (CD34 and Factor VIII staining) of 22-38% relative to controls depending on the specific tumor xenograft. The antitumor efficacy of chronic CEP-7055 administration was independent of initial tumor volume (in the ASPC-1 pancreatic carcinoma model) and reversible on withdrawal of treatment. Chronic p.o. administration of CEP-7055 in preclinical efficacy studies for periods of up to 65 days was well tolerated with no apparent toxicity or significant morbidity. Orally administered CEP-7055 has entered Phase I clinical trials in cancer patients.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Neoplasms, Experimental/drug therapy , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Administration, Oral , Angiogenesis Inhibitors/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Carbazoles/pharmacokinetics , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Female , In Vitro Techniques , Male , Mice , Mice, Nude , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/enzymology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/enzymology , Phosphorylation , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley , Solubility , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Xenograft Model Antitumor Assays
7.
Cancer Res ; 63(17): 5559-63, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14500395

ABSTRACT

All of the cases of medullary thyroid carcinoma (MTC) express the RET receptor tyrosine kinase. In essentially all of the hereditary cases and approximately 40% of the sporadic cases of MTC, the RET kinase is constitutively activated by mutation. This suggests that RET may be an effective therapeutic target for treatment of MTC. We show that the indolocarbazole derivatives, CEP-701 and CEP-751, inhibit RET in MTC cells. These compounds effectively inhibit RET phosphorylation in a dose-dependent manner at concentrations <100 nM in 0.5% serum and at somewhat higher concentrations in the presence of 16% serum. They also blocked the growth of these MTC cells in culture. CEP-751 and its prodrug, CEP-2563, also inhibited tumor growth in MTC cell xenografts. These results show that inhibiting RET can block the growth of MTC cells and may have a therapeutic benefit in MTC.


Subject(s)
Carbazoles/pharmacology , Carcinoma, Medullary/drug therapy , Indoles , Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Thyroid Neoplasms/drug therapy , Animals , Carcinoma, Medullary/enzymology , Carcinoma, Medullary/pathology , Cell Division/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Furans , Growth Inhibitors/pharmacology , Humans , Male , Mice , Mice, Nude , Multiple Endocrine Neoplasia Type 2b/genetics , Mutation , Oncogene Proteins/metabolism , Phosphorylation/drug effects , Prodrugs/pharmacology , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/metabolism , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Mol Cancer Ther ; 2(4): 371-82, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12700281

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP-1) is a nuclear zinc finger DNA-binding protein that is implicated in the repair of DNA damage. Inhibition of PARP-1 through genetic knockouts causes cells to become hypersensitive to various chemotherapeutic agents. We tested the chemopotentiating ability of the PARP-1 inhibitor, CEP-6800, when used in combination with temozolomide (TMZ), irinotecan (camptothecin or SN38), and cisplatin against U251MG glioblastoma, HT29 colon carcinoma, and Calu-6 non-small cell lung carcinoma xenografts and cell lines, respectively. Exposure of tumor cells to TMZ, camptothecin (or SN38), and cisplatin before, or in the presence of, CEP-6800 significantly increased the onset and the magnitude of DNA damage, the duration for cells to effect repair, and the onset, duration, or fraction of cells arrested at the G(2)/M boundary. In addition, in vivo biochemical efficacy studies with CEP-6800 showed that it was able to attenuate irinotecan- and TMZ-induced poly(ADP-ribose) accumulation in LoVo and HT29 xenografts, respectively. Treatment of CEP 6800 (30 mg/kg) with TMZ (17 and 34 mg/kg) resulted in 100% complete regression of U251MG tumors by day 28 versus 60% complete regression caused by TMZ alone. CEP-6800 (30 mg/kg) in combination with irinotecan (10 mg/kg) resulted in a 60% inhibition of HT29 tumor growth versus irinotecan alone by day 33. The combination therapy of cisplatin (5 mg/kg) with CEP-6800 (30 mg/kg) caused a 35% reduction in Calu-6 tumor growth versus cisplatin alone by day 28. These data suggest that CEP-6800 could be used as a chemopotentiating agent with a variety of clinically effective chemotherapeutic agents.


Subject(s)
Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cisplatin/pharmacology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle/drug effects , Cell Division , Cell Line , Cell Line, Tumor , DNA Damage , Dose-Response Relationship, Drug , Drug Synergism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Irinotecan , Kinetics , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Models, Chemical , Neoplasm Transplantation , Temozolomide , Time Factors
9.
Clin Cancer Res ; 8(6): 1924-31, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12060637

ABSTRACT

PURPOSE: Aberrant expression of trk receptor kinases and enhanced expression of various neurotrophins (NTs) have been implicated in the development and progression of human prostatic carcinoma and pancreatic ductal adenocarcinoma. We examined the antitumor efficacy of administration of NT neutralizing antibodies on the growth of established human prostatic carcinoma and pancreatic ductal adenocarcinoma xenografts in nude mice. EXPERIMENTAL DESIGN: In initial studies, tumor-bearing nude mice were treated with a mixture of NT antibodies [100 microg each of anti-nerve growth factor (NGF), anti-brain-derived neurotrophic factor, anti-NT-3, and anti-NT-4/5] or normal rabbit IgG (400 microg) intratumorally and peritumorally three times/week over a 15-day dosing period. In subsequent studies, tumor-bearing nude mice were treated with individual NT antibodies (100 microg), affinity-purified anti-NGF (0.1, 1.0, or 10.0 microg), or normal rabbit IgG (100 microg) using the same dosing schedule. RESULTS: Treatment with the antibody mixture inhibited significantly the growth of TSU-Pr1 and AsPC-1 xenografts as compared with IgG-treated controls (maximal inhibition of 53 and 53%, respectively), whereas this treatment caused significant regression in PC-3 xenografts. Treatment of TSU-Pr1 xenografts with either anti-NGF or anti-NT-3 resulted in maximal tumor growth inhibition of 67 and 64%, respectively, whereas anti-brain-derived neurotrophic factor and anti-NT-4/5 did not inhibit tumor growth in this tumor model. Administration of various concentrations (0.1, 1.0, or 10.0 microg) of affinity-purified anti-NGF resulted in maximal TSU-Pr1 tumor growth inhibition of 49, 62, and 66%, respectively. CONCLUSIONS: These data add further support for the therapeutic potential of disrupting trk-signaling events in select types of nonneuronal human cancers, specifically prostatic and pancreatic carcinomas.


Subject(s)
Carcinoma, Pancreatic Ductal/prevention & control , Nerve Growth Factors/physiology , Pancreatic Neoplasms/prevention & control , Prostatic Neoplasms/prevention & control , Receptor, trkA/physiology , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Division , Disease Progression , Female , Humans , Immunoglobulin G/therapeutic use , Male , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transplantation, Heterologous
10.
Blood ; 99(11): 3885-91, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12010785

ABSTRACT

Constitutively activating internal tandem duplication (ITD) and point mutations of the receptor tyrosine kinase FLT3 are present in up to 41% of patients with acute myeloid leukemia (AML). These FLT3/ITD mutations are likely to be important because their presence is associated with a poor prognosis. Both types of mutations appear to activate the tyrosine kinase activity of FLT3. We describe here the identification and characterization of the indolocarbazole derivative CEP-701 as a FLT3 inhibitor. This drug potently and selectively inhibits autophosphorylation of wild-type and constitutively activated mutant FLT3 in vitro in FLT3/ITD-transfected cells and in human FLT3-expressing myeloid leukemia-derived cell lines. We demonstrate that CEP-701 induces a cytotoxic effect on cells in a dose-responsive fashion that parallels the inhibition of FLT3. STAT5 and ERK1/2, downstream targets of FLT3 in the signaling pathway, are inhibited in response to FLT3 inhibition. In primary leukemia blasts from AML patients harboring FLT3/ITD mutations, FLT3 is also inhibited, with an associated cytotoxic response. Finally, using a mouse model of FLT3/ITD leukemia, we demonstrate that the drug inhibits FLT3 phosphorylation in vivo and prolongs survival. These findings form the basis for a planned clinical trial of CEP-701 in patients with AML harboring FLT3- activating mutations.


Subject(s)
Carbazoles/toxicity , Enzyme Inhibitors/pharmacology , Indoles , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Marrow Cells/enzymology , Bone Marrow Cells/pathology , Cell Survival/drug effects , Furans , Humans , Kinetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Point Mutation , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Cells, Cultured , fms-Like Tyrosine Kinase 3
SELECTION OF CITATIONS
SEARCH DETAIL
...